Вы находитесь на странице: 1из 4

TOXICITIES OF CALCINEURIN ANTAGONISTS

Chronic Cyclosporine Nephrotoxicity: A Pig Model


D. Cibulskyte, H. Kaalund, M. Pedersen, A. Hørlyck, N. Marcussen, H.E. Hansen, M. Madsen,
and J. Mortensen

ABSTRACT
Cyclosporine A (CsA) is one of the keystones in immunosuppressive treatment after solid
organ transplantation, despite its major side effects such as nephrotoxicity. The chronic
nephrotoxic effects of CsA seen in humans have been difficult to reproduce in small-animal
models. The aim of the present study was to examine the chronic nephrotoxicity produced
by therapeutic dosages of CsA in a pig model. Among 11 Gottingen minipigs included in
the study, three died, yielding data from five animals given CsA (10 mg/kg/d, orally) for 6
months, and three controls. Body weight, blood pressure, glomerular filtration rate (GFR)
by plasma clearance of 51Cr-ethylenediamine-tetraacetic acid, CsA concentration, serum
creatinine, and other values were measured every 5 weeks. Our results showed that the
whole blood trough CsA levels were lower in pigs than in humans treated with similar CsA
doses. Renal biopsies, which were obtained successfully, except one case of macroscopic
hematuria, showed no histological changes in the kidney. No significant increase in serum
creatinine or blood pressure was observed. Surprisingly, there was a significant increase in
GFR during CsA treatment. We conclude that the pig model displays a hyperfiltration that
warrants further investigation.

C YCLOSPORINE A (CsA) is an important immuno-


suppressive drug used in organ transplantation as well
as in the treatment of some primary renal and nonrenal
rotoxicity is the lack of a suitable animal model. CsA side
effects have been observed in a number of species, including
goat, sheep, baboon, dog, guinea pig, rabbit, and rat.10 The
immune-mediated diseases. CsA has led to a significant
reduction in renal allograft rejection, improving graft and
patient survival after its introduction for general use in the From the Departments of Renal Medicine (D.C., H.E.H., M.M.),
early 1980s.1–3 However, the drug has some serious side Magnetic Resonance (M.P.), and Radiology (A.H.), Skejby Syge-
effects, particularly chronic nephrotoxicity.4 –9 This condi- hus, Aarhus University Hospital, Aarhus, Denmark; Department
tion is manifested by renal insufficiency due to glomerular of Clinical Biochemistry (H.K.), Odense University Hospital,
and vascular changes, abnormalities of tubular function as Odense, Denmark; Department of Pathology (N.M.), Aarhus
Sygehus, Aarhus University Hospital, Aarhus, Denmark; and
well as hypertension. Renal biopsy reveals arteriolar hyli-
Institute of Experimental Clinical Research (J.M.), Skejby Syge-
nosis, focal interstial fibrosis, tubular atrophy, and glomer- hus, Aarhus University Hospital, Aarhus, Denmark.
ulosclerosis with focal atrophy. Address reprint requests to D. Cibulskyte, Department of
One of the reasons for the limited success of experimen- Renal Medicine, Aarhus University Hospital, Skejby Sygehus,
tal research studying the mechanism of chronic CsA neph- 8200 Aarhus N, Denmark. E-mail: donataci@yahoo.com

0041-1345/05/$–see front matter © 2005 by Elsevier Inc. All rights reserved.


doi:10.1016/j.transproceed.2005.09.004 360 Park Avenue South, New York, NY 10010-1710

3298 Transplantation Proceedings, 37, 3298 –3301 (2005)


CHRONIC CYCLOSPORINE NEPHROTOXICITY 3299

majority of studies using rodents as experimental models the bladder. Through a small incision the superficial femoral artery
have failed to show chronic CsA nephrotoxicity as seen in was cannulated for blood sampling and measurement of blood
humans treated with therapeutic doses.11 The structural pressure. At baseline (T0) and at 5-week intervals (T1 to T5), a
damage was only induced at CsA12–13 high doses or in battery of examinations was performed over 6 months.
The animals were weighed upon arrival. Blood samples were
salt-depleted rats, which have increased susceptibility to the
collected 18 hours after latest CsA dose for determination of serum
effects of CsA.14 In contrast, a study in Sprague-Dawley rats creatinine and of whole blood trough CsA concentrations using the
given 10 mg/kg of CsA intraperitoneally daily for 2 weeks EMIT 2000 Cyclosporin kit (Dade Bering). Blood pressure was
showed renal tubular dilatation, suggesting subtle tubular measured using a pressure transducer (Statham Gould #4523551)
damage and a precursor of focal interstitial fibrosis.15 The attached to a monitor (Medistim CardioMed CM-4008, Norway).
renal functional and histological changes have also been Glomerular filtration rate (GFR) was calculated by plasma clear-
produced in three other studies, where Sprague-Dawley ance of 51Cr-ethylenediamine-tetraacetic acid (EDTA) (Behring,
rats were administered 5 mg/kg/d, 12.5 mg/kg/d, or 15 Marburg, Germany). A bolus injection of 6 MBq 51Cr-EDTA was
mg/kg/d orally.16 –18 given intravenously followed by continuous infusion of 1.5 MBq
51
Experimental research on CsA nephrotoxicity has also Cr-EDTA/h during the experiment. After a loading period of 1
hour, a clearance period of 1 hour was used. The bladder was
been performed in rabbit models, but far less frequently
emptied at the beginning and at the end of the collection period.
than in the rat.11 One study in New Zealand rabbits used
Plasma 51Cr-EDTA was obtained at time 0 as well as at 20, 40, and
low doses of CsA (2.5, 5, or 10 mg/kg/d, intravenously), 60 minutes. The values of plasma and urine 51Cr-EDTA were
revealing morphological changes similar to those seen in estimated in a gamma counter (Cobra, Packard, USA) and cor-
man.19 The pig has been used to elucidate the mechanisms rected for radioactive decay. GFR was calculated as:
of the functional nephrotoxic effects of CsA.10 Vaden et al
GFR ⫽ UCr ⫻ V ⁄ PCr
produced mild multifocal renal tubular dilatation in York-
shire pigs, which received 25 mg/kg/d CsA orally for 22 where UCr is the concentration of 51Cr-EDTA in the collected
days.20 urine, V is the urine flow per unit of time, and PCr is the
The pig has several advantages as a model for pharma- interpolated plasma 51Cr-EDTA concentration of the four blood
cological studies,21 because of similar cardiovascular and samples.
Finally ultrasonography was performed to measure renal length
renal physiology and anatomy to man.22 From an economic
and gross morphology in addition to taking two kidney biopsies
point of view, it is obvious that the minipig is attractive from the lower pole using a Bard 18-G cannula. The frequency of
because of its low weight at birth, sexual maturity, and adult complications as urinary infection, bleeding and infection after
age. Ethically, such experiments cannot be performed in incision over the femoral artery and after renal biopsies was
man. However, there are some differences in the pharma- recorded. Upon completion the animals were awakened and taken
cokinetics of immunosuppressive drugs between pigs and back to the farm.
humans. Measuring CsA and prednisolone levels by high- At the final experiment a bilateral nephrectomy was performed.
pressure liquid chromatography after both intravenous and The animals were sacrificed using a lethal dose of potassium
oral doses, Frey et al23 reported that to obtain the same chloride. Kidney tissue and biopsies were examined for the follow-
target CsA concentrations, pigs required two and four to six ing histopathological changes: (1) arteriolar hyalinosis, (2) patchy
interstitial fibrosis, (3) glomerulosclerosis, and (4) tubular atrophy.
times higher doses, respectively. The aim of this work was to
The study protocol was approved by the Animal Ethics Council.
elucidate the nephrotoxic effects after 6 months of the CsA We used one-way repeated measures analysis of variance
dosage used after clinical kidney transplantation. (ANOVA) and Wilcoxon signed-rank test to assess changes within
the CsA-treated group. Comparisons between two groups were
MATERIALS AND METHODS performed using the Mann-Whitney test. A significance level of P
⬍ .05 was chosen.
Eleven Gottingen minipigs of body weight of 20 kg were housed in
individual cages on a farm and fed a standard pig diet. After the RESULTS
baseline evaluation, six animals were administered CsA (Sandim-
mun Neoral oral solution, Novartis Pharmaceuticals, Basel, Swit- Eleven Gottingen minipigs were included in the study: six
zerland; 10 mg/kg/d) in two divided oral doses. From the evening were given CsA and five were used as untreated controls.
before the investigations, the pigs abstained from food but had free Three animals died: one CsA-treated pig experienced re-
access to tap water. spiratory insufficiency due to a technical problem during
The animals were premedicated with azaperon (1 mg/10 kg) and anesthesia and two controls developed fulminate pulmo-
midazolam (1 mg/kg) intramuscularly. Anesthesia was induced by nary infections. Data were therefore obtained from five
etomidate (0.25 mg/kg intravenously) and maintained with keta- animals in the CsA group and three in the control group.
mine (6 mg/kg/h intravenously) and isofluran (0.5% to 0.75% in a
In all pigs two biopsies of sufficient size were obtained
volumetric mixture of O2 and N2O in a ratio of 1:1). The pigs were
without complication, except one case of macroscopic he-
orotracheally intubated and mechanically ventilated with a respi-
rator (Siemens, Servo 900D). Through an ear vein isotonic saline maturia for 2 hours. The femoral incision healed in all cases
alternating with 5% glucose (10 mL/kg/h) was given to maintain without complications.
hydration. Ampicillin (1 g intravenous) was delivered at both the The results of our study are shown in Table 1. From T0
beginning and the end of the investigations. For urine collection to T1 the CsA-treated pigs lost weight on average from
and measurement, a 12-F catheter was placed transurethrally into 20.5 to 18.3 kg (P ⬍ .03). Thereafter they gained weight
3300 CIBULSKYTE, KAALUND, PEDERSEN ET AL

Table 1. Functional and Chemical Parameters (Mean ⴞ SD) for the CsA-Treated Pigs Measured Baseline (T0) and Every
5 Weeks (T1–5) for 6 Months
Time Body weight (kg) GFR (mL/min) CsA (ng/mL) Creatinine (␮mol/L) Mean blood pressure (mm Hg)

T0 (0 wk) 20.2 ⫾ 1.3 48 ⫾ 10 90 ⫾ 10 79 ⫾ 18


T1 (5 wk) 17.8 ⫾ 1.4 44 ⫾ 15 81 ⫾ 35 68 ⫾ 10 75 ⫾ 18
T2 (10 wk) 18.1 ⫾ 2.1 87 ⫾ 25 27 ⫾ 8 65 ⫾ 16 77 ⫾ 12
T3 (15 wk) 19.8 ⫾ 2.6 113 ⫾ 76 49 ⫾ 24 94 ⫾ 29 81 ⫾ 14
T4 (20 wk) 21.6 ⫾ 2.9 67 ⫾ 16 53 ⫾ 40 83 ⫾ 8 82 ⫾ 8
T5 (25 wk) 23.1 ⫾ 3.1 100 ⫾ 27 64 ⫾ 57 78 ⫾ 15 80 ⫾ 10

for the rest of the experiment to an average weight of vessel changes, no patchy interstitial fibrosis, no glomeru-
23.0 kg. The increase from T1 to T5 was significant (P ⬍ losclerosis, and no tubular atrophy.
.05). The changes in weight from T1 to T5 in the
CsA-treated group were significantly different from the DISCUSSION
changes in the control group (P ⬍ .05), but there was also
As an experimental animal for the study of CsA-induced
a significant difference in weight between the two groups at
nephrotoxicity, the pig has advantages over the rat because
T0 (P ⬍ .05).
of its similarity to humans in cardiovascular as well as renal
Serum creatinine did not increase and mean blood
physiology and anatomy. Furthermore, rat studies do not
pressure was constant during the study (P ⫽ .940). GFR
permit serial biopsies. Previously, the rat has been popular
increased significantly during CsA administration (P ⫽
as an animal model because it is small, cheap, and has a fast
.031). The values were the same at T0 and T1 with a mean
turnover, so that studies can be performed quickly, which is
value of 46 mL/min. Subsequently, GFR increased steadily
one of the drawbacks of the pig. The Gottingen minipig,
for the rest of the experiment, reaching a value of 100
however, has an acceptable size for scientific investigations.
mL/min at T5.
A 10 mg/kg/d dosage of CsA is often used in clinical
Due to an insufficient number of 51Cr-EDTA clearance
practice after renal transplantation. We chose this dosage
results in the control group these data are omitted; we refer
for the experiment to observe effects during 6 months of
to the corresponding GFR estimates based on magnetic
treatment of CsA in an animal model reflecting humans.
resonance imaging (MRI) GFR data (Fig 1).24 The average
Whole blood trough concentrations of CsA were lower than
CsA whole blood trough concentration was 81 ng/mL at 5
in humans, due to differences in drug pharmacokinetics.
weeks and then varied around 50 ng/mL for the rest of the
The animals did not develop diarrhea. Little is known about
study period (Table 1). However, there were no significant
the dose of CsA, which can induce the nephrotoxic effects in
differences (P ⫽ .05). The kidney specimens showed no
the pig. In one study, pigs that received 25 mg/kg/d CsA
orally developed renal tubular dilatation, with blood 24-
hour trough concentrations measured by radio immunoas-
say being maintained at 641 ⫾ 175 ng/mL.20 Frey et al
found that the pig shows a large variability in the bioavail-
ability of CsA delivered by the oral route.23 The pig has also
a clearance rate that is considerably higher than that in
man. These findings may be attributable to different rates of
renal or biliary clearance, species-specific metabolic path-
ways, or differences in analytical methods.20
Surprisingly, the present study showed glomerular hyper-
filtration in the CsA-treated group starting between week 5
and 10. To our knowledge, this has not been described
previously during long-term CsA treatment; at present it is
unknown whether the observation is caused by CsA. In
diabetic nephropathy, renal function impairment is pre-
ceded by renal growth24 followed by hyperfiltration25 and
ultimately ending with deteriorating kidney function lead-
ing to end-stage renal failure. It is tempting to speculate
that the observed hyperfiltration found in this study may be
induced by CsA and might indicate an early phase of CsA
Fig 1. The relative GFR values for the CsA-treated group (grey nephrotoxicity. Further studies to evaluate these issues and
columns) and the control group (black columns) measured at the pharmacokinetics of CsA in the pig are in progress.
baseline (T0), 15 weeks (T3), 20 weeks (T4), and 25 weeks (T5). To conclude, our work indicates that the pig may be
CHRONIC CYCLOSPORINE NEPHROTOXICITY 3301

suitable as an animal model for studying the nephrotoxicity 13. Gillum DM, Truora L, Tasby J, et al: Chronic cyclosporine
of CsA. The increase in GFR during CsA treatment war- nephrotoxicity: a rodent model. Transplantation 46:285, 1988
rants further investigation. 14. Young BA, Burdmann EA, Johnson RJ, et al: Cyclosporine
A induced arteriolopathy in a rat model of chronic cyclosporine
nephrotoxicity. Kidney Int 48:431, 1995
REFERENCES 15. Ferguson CJ, von Ruhland C, Parry-Jones DJ, et al: Low
dose cyclosporine nephrotoxicity in the rat. Nephrol Dial Trans-
1. Kramer NC, Peters TG, Rohr MS, et al: Beneficial effects of plant 8:1259, 1993
cyclosporine on renal transplantation. Transplantation 49:343,
16. Backman L, Sundelin B, Bohman S-O: Focal glomeruloscle-
1990
rosis and nephron atrophy in rats on long-term cyclosporine
2. Opelz G: Collaborative transplant study—10-year report.
treatment. APMIS 4(suppl):27, 1988
Transplantation 24:2342, 1992
3. Danovitch GM: Choice of immunosuppressive drugs and 17. Dieperink H, Leyssac PP, Starklint H, et al: Long-term
individualization or immunosuppressive therapy for kidney trans- cyclosporin nephrotoxicity in the rat: effects on renal function and
plant patients. Transplant Proc 31(suppl 8A):2S, 1999 morphology. Nephrol Dial Transplant 3:317, 1988
4. de Mattos AM, Olyaei AJ, Bennett WM: Nephrotoxicity of 18. Sobh M, Sabry A, Moustafa F, et al: Effect of colchicines
immunosuppressive drugs: long-term consequences and challenges on chronic ciclosporin nephrotoxicity in Sprague-Dawley rats.
for the future. Am J Kidney Dis 35:333, 2000 Nephron 79:452, 1998
5. Olyaei AJ, de Mattos AM, Bennett WM: Nephrotoxicity of 19. Thliveris JA, Yatscoff RW, Lukowski MP, et al: Chronic
immunosuppressive drugs: new insight and preventive strategies. ciclosporine nephrotoxicity: a rabbit model. Nephron 57:470, 1991
Curr Opin Crit Care 7:384, 2001 20. Vaden SL, Riviere JE: Pharmacokinetics, inhibition of lym-
6. Wilkinson AH, Cohen DJ: Renal failure in the recipients of phoblast transformation, and toxicity of cyclosporine in clinically
nonrenal solid organ transplants. Am Soc Nephrol 10:1136, 1999 normal pigs. Am J Vet Res 51:399, 1990
7. Olyaei AJ, de Mattos AM, Bennett WM: Immunosuppressant- 21. Bollen P, Ellegaard L: The Gottingen minipig in pharmacol-
induced nephropathy: pathophysiology, incidence and management. ogy and toxicology. Pharmacol Toxicol 80(suppl 2):3, 1997
Drug Safety 21:471, 1999
22. Terris JM: Swine as a model in renal physiology and
8. Campistol JM, Sacks SH: Mechanisms of nephrotoxicity.
nephrology: an overview. Swine in biomedical research. Vol. 3.
Transplantation 69(suppl):SS5, 2000
New York and London: Plenum Press; 1986, p 1673
9. Bennett WM, de Mattos A, Meyer MM, et al: Chronic
cyclosporine nephropathy: the Achilles’ heel of immunosuppres- 23. Frey BM, Sieber M, Mettker D, et al: Marked interspecies
sive therapy. Kidney Int 50:1089, 1996 differences between humans and pigs in cyclosporine and pred-
10. Calvert DA, Terris JM: Evaluation of the mechanism of nisolone disposition. Drug Metabolism and Disposition 16:285,
cyclosporine A nephrotoxicity. Advances in swine in biomedical 1988
research. Vol 1. New York: Plenum Press; 1996, p 67 24. Khristiansen T, Stoedkilde-Jorgensen H, Flyvbjerg A: Mag-
11. Thliveris JA, Yatscoff RW, Lukowski MP, et al: Cyclospor- netic resonance imaging as a monitor of kidney enlargement and
ine nephrotoxicity— experimental models. Clin Biochem 24:93, regression in experimental diabetes in rats. J Endocrinol 153:193,
1991 1997
12. Jackson NM, Hsu C-H, Visscher GE, et al: Alternations in 25. Bak M, Thomsen K, Christiansen T, et al: Renal enlarge-
renal structure and function in a rat model of cyclosporine neph- ment precedes renal hyperfiltration in early diabetes in rats. J Am
rotoxicity. J Pharmacol Exp Ther 242:749, 1987 Soc Nephrol 11:1287, 2000

Вам также может понравиться